Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Clin Res Hepatol Gastroenterol ; 48(4): 102318, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38471582

ABSTRACT

BACKGROUND: Concurrent chemo-radiotherapy (CCRT) is the preferred non-surgical treatment for patients with locally advanced esophageal squamous cell carcinoma (ESCC). Unfortunately, some patients respond poorly, which leads to inappropriate or excessive treatment and affects patient survival. To accurately predict the response of ESCC patients to CCRT, we developed classification models based on the clinical, serum proteomic and radiomic data. METHODS: A total of 138 ESCC patients receiving CCRT were enrolled in this study and randomly split into a training cohort (n = 92) and a test cohort (n = 46). All patients were classified into either complete response (CR) or incomplete response (non-CR) groups according to RECIST1.1. Radiomic features were extracted by 3Dslicer. Serum proteomic data was obtained by Olink proximity extension assay. The logistic regression model with elastic-net penalty and the R-package "rms" v6.2-0 were applied to construct classification and nomogram models, respectively. The area under the receiver operating characteristic curves (AUC) was used to evaluate the predictive performance of the models. RESULTS: Seven classification models based on multi-omics data were constructed, of which Model-COR, which integrates five clinical, five serum proteomic, and seven radiomic features, achieved the best predictive performance on the test cohort (AUC = 0.8357, 95 % CI: 0.7158-0.9556). Meanwhile, patients predicted to be CR by Model-COR showed significantly longer overall survival than those predicted to be non-CR in both cohorts (Log-rank P = 0.0014 and 0.027, respectively). Furthermore, two nomogram models based on multi-omics data also performed well in predicting response to CCRT (AUC = 0.8398 and 0.8483, respectively). CONCLUSION: We developed and validated a multi-omics based classification model and two nomogram models for predicting the response of ESCC patients to CCRT, which achieved the best prediction performance by integrating clinical, serum Olink proteomic, and radiomic data. These models could be useful for personalized treatment decisions and more precise clinical radiotherapy and chemotherapy for ESCC patients.


Subject(s)
Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , Humans , Esophageal Neoplasms/therapy , Esophageal Squamous Cell Carcinoma/therapy , Multiomics , Proteomics , Pathologic Complete Response , Chemoradiotherapy , Retrospective Studies
2.
Oncogene ; 42(30): 2315-2328, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37353616

ABSTRACT

SOX2 is highly expressed and controls tumor initiation and cancer stem cell function in various squamous cell carcinomas including esophageal squamous cancer. However, the molecular mechanism leading to SOX2 overexpression in cancer is incompletely understood. Here, we identified CHIP, a chaperone-associated ubiquitin E3 ligase, as a novel negative regulator of SOX2 protein stability and tumorigenic activity in esophageal squamous carcinoma cells. We showed that CHIP interacted with SOX2 primarily via chaperone HSP70, together they catalyzed SOX2 ubiquitination and degradation via proteasome. In contrast, HSP90 promoted SOX2 stability and inhibition of HSP90 activity induced SOX2 ubiquitination and degradation. Notably, unlike the case in normal esophageal tissues where CHIP was detected in both the cytoplasm and nucleus, CHIP in clinical esophageal tumor specimens was predominantly localized in the cytoplasm. Consistent with this observation, we observed increased expression of exportin-1/CRM-1 in clinical esophageal tumor specimens. We further demonstrated that CHIP catalyzed SOX2 ubiquitination and degradation primarily in the nuclear compartment. Taken together, our study has identified CHIP as a key suppressor of SOX2 protein stability and tumorigenic activity and revealed CHIP nuclear exclusion as a potential mechanism for aberrant SOX2 overexpression in esophageal cancer. Our study also suggests HSP90 inhibitors as potential therapeutic agents for SOX2-positive cancers.


Subject(s)
Esophageal Neoplasms , Ubiquitin-Protein Ligases , Humans , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , Proteasome Endopeptidase Complex/metabolism , Molecular Chaperones/metabolism , Esophageal Neoplasms/genetics , Protein Stability , SOXB1 Transcription Factors/metabolism
3.
Oncogene ; 42(30): 2297-2314, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37349645

ABSTRACT

Esophageal squamous cell carcinoma (ESCC) is one of the deadliest forms of human malignancy that currently lacks approved targeted therapeutics. Accumulating evidence suggests that SOX2 overexpression is a key driving factor for ESCC and various squamous cell carcinoma. Here, through screening a small-molecule kinase inhibitor library, we identified GSK3ß as a kinase that is critically required for robust SOX2 expression in ESCC cells. GSK3ß did not promote SOX2 transcriptionally but was required for SOX2 protein stability. We demonstrated that GSK3ß interacts with and phosphorylates SOX2 at residue S251, which blocks SOX2 from ubiquitination and proteasome-dependent degradation instigated by ubiquitin E3 ligase CUL4ADET1-COP1. Pharmacological inhibition or knockdown of GSK3ß by RNA interference selectively impaired SOX2-positive ESCC cell proliferation, cancer stemness, and tumor growth in mouse xenograft model, suggesting that GSK3ß promotes ESCC tumorigenesis primarily by driving SOX2 overexpression. GSK3ß was found to be frequently overexpressed in clinical esophageal tumors, and there was a positive correlation between GSK3ß and SOX2 protein levels. Notably, we found that SOX2 enhanced GSK3ß expression transcriptionally, suggesting the existence of a vicious cycle that drives a coordinated GSK3ß and SOX2 overexpression in ESCC cells. Finally, we demonstrated in tumor xenograft model that GSK3ß inhibitor AR-A014418 was effective in suppressing SOX2-positive ESCC tumor progression and inhibited tumor progression cooperatively with chemotherapeutic agent carboplatin. In conclusion, we uncovered a novel role for GSK3ß in driving SOX2 overexpression and tumorigenesis and provided evidence that targeting GSK3ß may hold promise for the treatment of recalcitrant ESCCs.


Subject(s)
Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , Humans , Animals , Mice , Esophageal Squamous Cell Carcinoma/drug therapy , Esophageal Neoplasms/pathology , Glycogen Synthase Kinase 3 beta/genetics , Glycogen Synthase Kinase 3 beta/metabolism , Cell Line, Tumor , Carcinogenesis/genetics , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , Cullin Proteins/genetics , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism
4.
Cell Death Differ ; 30(2): 527-543, 2023 02.
Article in English | MEDLINE | ID: mdl-36526897

ABSTRACT

Anillin (ANLN) is a mitosis-related protein that promotes contractile ring formation and cytokinesis, but its cell cycle-dependent degradation mechanisms in cancer cells remain unclear. Here, we show that high expression of ANLN promotes cytokinesis and proliferation in esophageal squamous cell carcinoma (ESCC) cells and is associated with poor prognosis in ESCC patients. Furthermore, the findings of the study showed that the deubiquitinating enzyme USP10 interacts with ANLN and positively regulates ANLN protein levels. USP10 removes the K11- and K63-linked ubiquitin chains of ANLN through its deubiquitinase activity and prevents ANLN ubiquitin-mediated degradation. Importantly, USP10 promotes contractile ring assembly at the cytokinetic furrow as well as cytokinesis by stabilizing ANLN. Interestingly, USP10 and the E3 ubiquitin ligase APC/C co-activator Cdh1 formed a functional complex with ANLN in a non-competitive manner to balance ANLN protein levels. In addition, the macrolide compound FW-04-806 (F806), a natural compound with potential for treating ESCC, inhibited the mitosis of ESCC cells by targeting USP10 and promoting ANLN degradation. F806 selectively targeted USP10 and inhibited its catalytic activity but did not affect the binding of Cdh1 to ANLN and alters the balance of the USP10-Cdh1-ANLN complex. Additionally, USP10 expression was positively correlated with ANLN level and poor prognosis of ESCC patients. Overall, targeting the USP10-ANLN axis can effectively inhibit ESCC cell-cycle progression.


Subject(s)
Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , Humans , Esophageal Squamous Cell Carcinoma/genetics , Esophageal Neoplasms/metabolism , Contractile Proteins/metabolism , Ubiquitin/metabolism , Cell Proliferation , Cell Line, Tumor , Ubiquitin Thiolesterase/genetics , Ubiquitin Thiolesterase/metabolism
5.
Cell Death Dis ; 13(5): 496, 2022 05 25.
Article in English | MEDLINE | ID: mdl-35614034

ABSTRACT

Esophageal squamous cell carcinoma (ESCC) is one of the world's leading causes of death, and its primary clinical therapy relies on surgical resection, chemotherapy, radiotherapy, and chemoradiotherapy. Although the genomic features and clinical significance of ESCC have been identified, the outcomes of targeted therapies are still unsatisfactory. Here, we demonstrate that mitogen-activated protein kinase (MAPK) signaling is highly activated and associated with poor prognosis in patients with ESCC. Mitogen-activated protein kinase kinase (MEK) inhibitors efficiently blocked the phosphorylation of extracellular signal-regulated kinase 1/2 (ERK1/2) in ESCC, while signal transducer and activator of transcription 3 (STAT3) signaling was rapidly activated. Combined STAT3 inhibition prevented the emergence of resistance and enhanced MEK inhibitor-induced cell cycle arrest and senescence in vitro and in vivo. Mechanistic studies revealed that the suppressor of cytokine signaling 3 (SOCS3) was downregulated, resulting in an increase in STAT3 phosphorylation in MEK-inhibited cells. Furthermore, chromatin immunoprecipitation showed that ELK1, which was activated by MEK/ERK signaling, induced SOCS3 transcription. These data suggest that the development of combined MEK and STAT3 inhibition could be a useful strategy in ESCC targeted therapy.


Subject(s)
Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , MAP Kinase Signaling System , Mitogen-Activated Protein Kinase Kinases , Protein Kinase Inhibitors , STAT3 Transcription Factor , Cell Line, Tumor , Esophageal Neoplasms/drug therapy , Esophageal Neoplasms/genetics , Esophageal Neoplasms/metabolism , Esophageal Squamous Cell Carcinoma/drug therapy , Esophageal Squamous Cell Carcinoma/genetics , Esophageal Squamous Cell Carcinoma/metabolism , Humans , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinase Kinases/metabolism , Protein Kinase Inhibitors/pharmacology , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Suppressor of Cytokine Signaling Proteins/metabolism
6.
Cancer Lett ; 522: 171-183, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34571081

ABSTRACT

The clinical efficacy of cisplatin in the treatment of esophageal squamous cell carcinoma (ESCC) is undesirable. Signal transducer and activator of transcription 3ß (STAT3ß), a splice variant of STAT3, restrains STAT3α activity and enhances chemosensitivity in ESCC. However, the underlying molecular mechanisms remain poorly understood. Here, we found that high expression of STAT3ß contributes to cisplatin sensitivity and enhances Gasdermin E (GSDME) dependent pyroptosis in ESCC cells after exposure to cisplatin. Mechanistically, STAT3ß was located into the mitochondria and its high expression disrupts the activity of the electron transport chain, resulting in an increase of ROS in cisplatin treatment cells. While high levels of ROS caused activation of caspase-3 and GSDME, and induced cell pyroptosis. STAT3ß blocked the phosphorylation of STAT3α S727 in mitochondria by interacting with ERK1/2 following cisplatin treatment, disrupting electron transport chain and inducing activation of GSDME. Clinically, high expression of both STAT3ß and GSDME was strongly associated with better overall survival and disease-free survival of ESCC patients. Overall, our study reveals that STAT3ß sensitizes ESCC cells to cisplatin by disrupting mitochondrial electron transport chain and enhancing pyroptosis, which demonstrates the prognostic significance of STAT3ß in ESCC therapy.


Subject(s)
Caspase 3/genetics , Esophageal Squamous Cell Carcinoma/drug therapy , Receptors, Estrogen/genetics , STAT3 Transcription Factor/genetics , Cell Line, Tumor , Cisplatin/pharmacology , Drug Resistance, Neoplasm/genetics , Electron Transport/genetics , Esophageal Squamous Cell Carcinoma/genetics , Esophageal Squamous Cell Carcinoma/pathology , Female , Humans , Male , Mitochondria/drug effects , Mitochondria/genetics , Phosphorylation/drug effects , Pyroptosis/drug effects
7.
Curr Med Chem ; 28(19): 3787-3802, 2021.
Article in English | MEDLINE | ID: mdl-33121398

ABSTRACT

Cyclophilin A (CypA) is a ubiquitous and highly conserved protein. CypA, the intracellular target protein for the immunosuppressant cyclosporine A (CsA), plays important cellular roles through peptidyl-prolyl cis-trans isomerase (PPIase). Increasing evidence shows that CypA is up-regulated in a variety of human cancers. In addition to being involved in the occurrence and development of multiple tumors, overexpression of CypA has also been shown to be strongly associated with malignant transformation. Surgery, chemotherapy and radiotherapy are the three main treatments for cancer. Chemotherapy and radiotherapy are often used as direct or adjuvant treatments for cancer. However, various side effects and resistance to both chemotherapy and radiotherapy bring great challenges to these two forms of treatment. According to recent reports, CypA can improve the chemosensitivity and/or radiosensitivity of cancers, possibly by affecting the expression of drug-resistant related proteins, cell cycle arrest and activation of the mitogen-activated protein kinase (MAPK) signaling pathways. In this review, we focus on the role of CypA in cancer, its impact on cancer chemotherapeutic and radiotherapy sensitivity, and the mechanism of action. It is suggested that CypA may be a novel potential therapeutic target for cancer chemotherapy and/or radiotherapy.


Subject(s)
Cyclophilin A , Neoplasms , Cyclosporine , Humans , Immunosuppressive Agents , Peptidylprolyl Isomerase
9.
Pathology ; 41(7): 681-6, 2009.
Article in English | MEDLINE | ID: mdl-20001348

ABSTRACT

AIMS: The aim of this study was to characterise clinical and microbiological features of isolates obtained from both invasive and non-invasive Streptococcus pyogenes infections in Hong Kong, between October 2005 and April 2008. METHOD: Clinical data of invasive isolates were collected retrospectively. Altogether 281 isolates were emm sequence typed and tested for antimicrobial susceptibility using disk diffusion method. Detection of the presence of the streptococcal pyrogenic exotoxin genes was also carried out. RESULTS: emm1, emm4 and emm12 were the most prevalent in both the invasive and non-invasive groups with an increase in incidence of emm22 compared with a previous study. emm22 was associated with invasive cellulitis and wound infection. The overall rate of erythromycin resistance was 25.6% and was significantly higher in emm22 strains (85.7%). The phage-encoded superantigen gene speA was exclusively associated with emm1 in both invasive and non-invasive isolates. CONCLUSION: This study revealed a changing epidemiology of S. pyogenes infection in Hong Kong, with a unique pattern compared with other Asian countries. Invasiveness is not related to the presence of speA, speC or ssa genes and the antimicrobial resistance rate was high for macrolides. The findings have an implication on the use and efficacy of the polyvalent S. pyogenes vaccine under development.


Subject(s)
Drug Resistance, Microbial/genetics , Streptococcal Infections/epidemiology , Streptococcal Infections/microbiology , Streptococcus pyogenes/isolation & purification , Anti-Bacterial Agents/pharmacology , Bacterial Outer Membrane Proteins/genetics , Bacterial Proteins/genetics , Drug Resistance, Microbial/drug effects , Erythromycin/pharmacology , Exotoxins/genetics , Genes, Bacterial/genetics , Hong Kong/epidemiology , Humans , Membrane Proteins/genetics , Microbial Sensitivity Tests , Molecular Epidemiology , Retrospective Studies , Streptococcal Infections/blood , Streptococcus pyogenes/genetics , Superantigens/genetics
10.
J Clin Microbiol ; 42(7): 3321-3, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15243105

ABSTRACT

Pulsed-field gel electrophoresis fingerprints of 98 Campylobacter jejuni isolates from patients (85) and chicken carcasses (13) in Hong Kong in 2002 demonstrated high genetic diversity. The prevalence of quinolone resistance among the isolates was 85.9%, and replacement of the threonine-86 residue in the gyrase subunit A was the major resistance mechanism.


Subject(s)
Anti-Infective Agents/pharmacology , Campylobacter jejuni/drug effects , Ciprofloxacin/pharmacology , Animals , Base Sequence , Campylobacter jejuni/genetics , Chickens/microbiology , Drug Resistance, Bacterial , Electrophoresis, Gel, Pulsed-Field , Humans , Microbial Sensitivity Tests , Molecular Sequence Data
SELECTION OF CITATIONS
SEARCH DETAIL
...